Germline CTLA-4 KO mice exhibit fatal lymphoproliferative disorders, with death occurring by 3–5 weeks due to multiorgan infiltration by hyperactivated T cells . Key findings include:
Lymphoproliferation: Massive expansion of CD4+ T cells and Foxp3+ regulatory T cells (Tregs), leading to splenomegaly and lymphadenopathy .
Autoimmunity: Organ-specific pathologies include pancreatitis, myocarditis, and insulitis, mimicking human autoimmune conditions like Sjögren’s syndrome .
Disease Modulation: Congenital KO mice develop myocarditis, while adult-inducible KO models (via tamoxifen-activated Cre recombinase) show delayed autoimmune onset without fatal pancreatitis .
Feature | Congenital KO | Inducible KO |
---|---|---|
Survival | ≤21 days | >100 days |
Key Pathology | Myocarditis, pancreatitis | Pneumonitis, sialadenitis |
Treg Expansion | Severe | Moderate |
Autoantibody Production | Pan-reactive | Organ-specific |
To bridge translational gaps, human CTLA-4 knock-in mice (huCTLA-4) express human extracellular/transmembrane domains while retaining murine cytoplasmic regions . These models enable:
Therapeutic Testing: Evaluation of anti-CTLA-4 antibodies (e.g., ipilimumab) in immuno-oncology .
Functional Conservation: Human CTLA-4 binds murine B7-1/B7-2, preserving ligand interactions .
Disease Studies: huCTLA-4 mice develop autoimmune-like conditions when crossed with susceptible strains, aiding studies on checkpoint inhibitor toxicity .
Conditional KO systems (e.g., Rosa26-Cre/ERT2) allow temporal control of CTLA-4 deletion:
Treg-Specific Deletion: Causes spontaneous autoimmunity, highlighting CTLA-4’s role in Treg-mediated suppression .
B-Cell-Specific Effects: CTLA-4 loss in B-1a cells increases autoantibodies and germinal center formation .
The CHO-K1/mouse CTLA-4 cell line (GenScript M00570) facilitates antibody screening and binding assays:
Surface Expression: Validated via FACS using PE anti-mouse CD152 .
Applications: Used for quantifying antibody affinity and functional blocking studies .
Key reagents include Bio X Cell’s InVivoMAb anti-mouse CTLA-4 (Clone 9D9):
Mechanism: Blocks CTLA-4/B7 interactions, enhancing T-cell responses .
Applications: Tumor immunotherapy studies and autoimmune disease modeling .
Central vs. Peripheral Tolerance: Congenital CTLA-4 KO disrupts thymic selection, while inducible KO impairs peripheral Treg function .
Model-Specific Outcomes: CTLA-4 deletion exacerbates collagen-induced arthritis but protects against peptide-induced experimental autoimmune encephalomyelitis (EAE) .
Checkpoint Blockade: Anti-CTLA-4 antibodies amplify antitumor immunity but risk autoimmune adverse effects .
Soluble CTLA-4: Mouse models reveal its role in enhancing Treg suppressive activity, linked to type 1 diabetes .
Emerging models like genO-hCTLA-4 mice (genOway) enable long-term studies of human CTLA-4-targeting therapies in immunocompetent hosts . These systems address cross-reactivity challenges and improve preclinical predictability.
CTLA-4 knockout mice develop a severe lymphoproliferative disorder characterized by massive expansion of lymphocytes, infiltration and destruction of major organs. These mice typically die at 2-3 weeks of age due to this aggressive autoimmune-like condition. The T cells in these mice display hyperactivation markers including CD69 and IL-2R expression, down-regulated CD62L, and spontaneous proliferation in vitro. Additionally, CD4+ T cells from these mice secrete elevated levels of IL-4 and IL-5 and demonstrate increased survival correlated with higher Bcl-xL expression . This lethal phenotype underscores CTLA-4's fundamental role in maintaining immune homeostasis.
Conditional deletion of CTLA-4 in adult mice results in a distinct and less severe phenotype compared to germline knockouts. While adult CTLA-4 deletion still causes lymphoproliferation, hypergammaglobulinemia, and organ-specific autoimmunity (including pneumonitis, gastritis, insulitis, and sialadenitis), this condition is not fatal unlike the congenital deficiency. Interestingly, conditional CTLA-4 deletion leads to preferential expansion of CD4+Foxp3+ regulatory T cells, though these T cells can still transfer disease into T cell-deficient mice . Additionally, the tissue targets of autoimmunity differ between congenital and adult CTLA-4 deficiency, with myocarditis uniquely developing after adoptive transfer of thymocytes from mice with congenital deficiency but not observed in donor mice depleted in adulthood .
The genO-hCTLA-4 mouse model has the murine CTLA-4 replaced with the human version, allowing for testing of human-specific CTLA-4-targeting therapeutics. These mice express different human CTLA-4 isoforms with physiological regulation and expression patterns while maintaining a fully functional mouse immune system. This model enables assessment of efficacy, mechanism of action, safety, and toxicity of CTLA-4-targeting compounds in the context of a complete immune system. It's particularly valuable for tumor growth inhibition assays in monotherapies or combination therapies, and supports long-term studies as immunological memory is observed following treatment .
Researchers can manipulate CTLA-4 expression through several approaches:
Germline knockout: Complete deletion of CTLA-4 from conception, resulting in early lethality
Conditional deletion: Using Cre-loxP systems to delete CTLA-4 in adult mice, enabling study of CTLA-4 function beyond early development
Humanized knock-in: Replacing mouse CTLA-4 exons (particularly exons 2 and 3 encoding extracellular and transmembrane domains) with human counterparts while maintaining the conserved cytoplasmic domain
Pharmacological targeting: Using antibodies that either block CTLA-4 function or deplete CTLA-4-expressing cells
For creating humanized CTLA-4 mice, researchers typically employ homologous recombination to replace mouse exons 2 and 3 with human equivalents. RT-PCR verification using primers spanning mouse exons 1-4 (648 bp product) and primers specific for mouse and human CTLA-4 can confirm proper expression and splicing .
Effective methods for assessing CTLA-4 function include:
T cell phenotyping: Flow cytometry to assess activation markers (CD69, IL-2R), lymphoid homing receptors (CD62L), and regulatory T cell markers (Foxp3)
Proliferation assays: Measuring spontaneous proliferation and response to costimulatory signals like anti-CD28
Cytokine profiling: Quantifying IL-4, IL-5, and IL-17 production by T cell subsets
Survival assessments: Evaluating Bcl-xL expression and T cell longevity in culture
Histopathological analysis: Examining tissue infiltration and damage in organs (lungs, stomach, pancreas, salivary glands)
Autoantibody detection: Measuring organ-specific autoantibodies
Adoptive transfer experiments: Transferring T cells into T cell-deficient recipients to assess disease induction potential
In one methodological approach, stimulating cells with anti-CD3 (2C11) for 30 hours allows for RNA extraction and RT-PCR amplification of CTLA-4 sequences to verify expression levels .
The relationship between anti-CTLA-4 antibody-induced tumor immunity and autoimmunity is not strictly correlated, contrary to initial assumptions. Studies using human CTLA-4 knock-in mice have shown that some antibodies can induce strong protection against cancer while causing minimal autoimmune side effects . The mechanisms behind this differential effect involve:
Distinct mechanisms of action in different tissues: CTLA-4 blockade may preferentially affect tumor-infiltrating Tregs versus peripheral Tregs
Antibody isotype effects: Different Fc regions can determine whether antibodies deplete CTLA-4-expressing cells through ADCC (antibody-dependent cellular cytotoxicity) and ADCP (antibody-dependent cellular phagocytosis) by NK cells and macrophages
Tumor microenvironment factors: The tumor microenvironment may alter CTLA-4 expression patterns and function
Importantly, studies with ipilimumab (an IgG1 anti-CTLA-4 antibody) suggest it may act primarily by depleting regulatory T cells within the tumor bed through ADCC and ADCP mechanisms rather than by simply blocking CTLA-4 signaling .
Several approaches show promise for reducing autoimmune side effects while preserving anti-tumor efficacy:
Fc engineering: Modifying the Fc regions of anti-CTLA-4 antibodies to alter their interaction with Fc receptors on NK cells and macrophages
Selective targeting: Developing antibodies that preferentially bind to specific CTLA-4 epitopes or conformations that may be enriched in tumor environments
Combinatorial approaches: Using lower doses of CTLA-4 antibodies in combination with other checkpoint inhibitors
CTLA-4 signaling peptides: Novel approaches like dNP2-ctCTLA-4 peptide that can induce regulatory T cells while inhibiting IL-17 production from T cells have shown efficacy in reducing inflammation without completely blocking CTLA-4 function
A comparative study showed that dNP2-ctCTLA-4 peptide ameliorated psoriasis progression through increasing Treg cells and inhibiting IL-17 production from γδ T cells, while CTLA-4-Ig did not show the same effect. This highlights the potential of novel CTLA-4-targeting strategies that preserve regulatory functions while mitigating inflammation .
CTLA-4 deficiency has divergent effects on different autoimmune disease models:
Disease Model | Effect of CTLA-4 Deficiency | Mechanism |
---|---|---|
Collagen-induced arthritis | Exacerbated disease | Enhanced T cell activation and reduced regulatory function |
Peptide-induced EAE | Protection | Altered T cell polarization or trafficking |
Protein-induced EAE | Delayed onset but not protection | Different antigen processing requirements |
Psoriasis | Exacerbated disease | Reduced Treg/Th17 ratio |
When working with CTLA-4 humanized mice, researchers should implement these critical controls:
Wild-type littermates: Essential for establishing baseline immune parameters
Heterozygous controls: To assess gene dosage effects (human/mouse CTLA-4 heterozygous mice)
Isotype control antibodies: For anti-CTLA-4 antibody studies
Expression verification: Using RT-PCR with primers specific for mouse and human CTLA-4 sequences
Functional verification: Confirming human CTLA-4 binding to mouse B7-1 and B7-2
Protein expression analysis: Flow cytometry using both anti-mouse and anti-human CTLA-4 antibodies
In humanized CTLA-4 studies, diagonal distribution of human and mouse CTLA-4 molecules in heterozygous mice reveals that both alleles are regulated by the same mechanisms, confirming proper physiological regulation of the human gene .
When faced with conflicting results between different CTLA-4 mouse models, researchers should consider:
Developmental timing: Results from germline knockout versus conditional deletion models may differ due to developmental compensation mechanisms
Genetic background: Different mouse strains may show variable susceptibility to autoimmunity
Environmental factors: Microbiome composition and pathogen exposure can significantly influence immune phenotypes
Model-specific mechanisms: Some models may primarily affect CTLA-4 signaling while others affect protein expression or trafficking
Completeness of deletion/blockade: Residual CTLA-4 expression or function may account for phenotypic differences
For instance, the less severe pro-autoimmune effect observed with some anti-CTLA-4 antibodies compared to others may result from subtle differences in their interaction with CTLA-4, affecting intracellular signaling or interactions with binding partners like B7-1 and B7-2 .
CTLA-4 mouse models are instrumental in studying combination immunotherapies through:
Dual checkpoint blockade: Combining CTLA-4 inhibition with PD-1/PD-L1 blockade to assess synergistic effects
Sequential therapy approaches: Testing optimal timing of CTLA-4 targeting relative to other immunotherapies
Combination with conventional therapies: Evaluating CTLA-4 blockade with chemotherapy, radiation, or targeted therapies
Biomarker identification: Determining predictive biomarkers for response to combination therapies
Novel combination partners: Testing emerging immunomodulatory agents in combination with CTLA-4 targeting
The genO-hCTLA-4 mouse model specifically enables assessment of human-specific antibodies in realistic immunological contexts, allowing for both monotherapy and combination therapy tumor growth inhibition assays .
CTLA-4's role in regulatory T cells extends beyond simple inhibition and includes:
Cell-extrinsic suppression: CTLA-4 on Tregs can capture and remove B7 molecules from antigen-presenting cells through transendocytosis
Metabolic regulation: CTLA-4 signaling influences cellular metabolism in Tregs, affecting their survival and function
Cytokine modulation: CTLA-4 engagement alters cytokine production and response profiles
Tissue-specific functions: CTLA-4 may have different roles in lymphoid versus non-lymphoid tissues
Developmental programming: CTLA-4 influences Treg development and stability
Studies using dNP2-ctCTLA-4 peptide demonstrated that enhancing CTLA-4 signaling increased Treg cell proportion while reducing IL-17 production by T cells, highlighting CTLA-4's role in maintaining immune homeostasis through Treg promotion .
Cytotoxic T-Lymphocyte Associated Antigen-4 (CTLA-4), also known as CD152, is a critical immune checkpoint receptor that plays a significant role in regulating immune responses. It is predominantly expressed on the surface of activated T cells and regulatory T cells (Tregs). The recombinant form of CTLA-4, particularly from mouse models, has been extensively studied to understand its biological functions and therapeutic potential.
CTLA-4 is a single-chain transmembrane glycoprotein belonging to the immunoglobulin superfamily. In mice, the CTLA-4 protein consists of 162 amino acids with a molecular weight ranging between 25-30 kDa . It is expressed mainly on CD4+ T cells and binds to the B7 family molecules (CD80 and CD86) on antigen-presenting cells with higher affinity than its homolog, CD28 .
CTLA-4 serves as a negative regulator of T cell activation. Upon binding to its ligands, CTLA-4 transmits inhibitory signals that dampen T cell responses, thereby maintaining immune homeostasis and preventing autoimmunity . This inhibitory function is crucial for the survival and function of Tregs, which play a pivotal role in suppressing immune responses and maintaining self-tolerance .
CTLA-4’s role extends to tumor immunity, where it modulates T cell responses to aid tumor cells in evading immune detection . By inhibiting T cell activation and proliferation, CTLA-4 contributes to the immunosuppressive tumor microenvironment. This has led to the development of anti-CTLA-4 antibody therapies aimed at blocking CTLA-4 to restore T cell activation and enhance anti-tumor immunity .
The therapeutic potential of targeting CTLA-4 has been explored in various clinical settings. Monoclonal antibodies against CTLA-4, such as ipilimumab, have shown promising results in treating several malignancies, including melanoma, renal cell carcinoma, and non-small cell lung carcinoma . These therapies work by blocking CTLA-4, thereby reversing Treg-mediated suppression and promoting robust anti-tumor immune responses .
Recombinant mouse CTLA-4 proteins are widely used in research to study the molecular mechanisms of CTLA-4-mediated immune regulation. These studies have provided valuable insights into the role of CTLA-4 in immune homeostasis, autoimmunity, and cancer . Ongoing research aims to further optimize CTLA-4 blockade and explore its potential in combination with other immune checkpoint inhibitors for enhanced therapeutic efficacy .